Protein Binders and Their Applications in Developmental Biology Stefan Harmansa* and Markus Affolter‡

Total Page:16

File Type:pdf, Size:1020Kb

Protein Binders and Their Applications in Developmental Biology Stefan Harmansa* and Markus Affolter‡ © 2018. Published by The Company of Biologists Ltd | Development (2018) 145, dev148874. doi:10.1242/dev.148874 PRIMER Protein binders and their applications in developmental biology Stefan Harmansa* and Markus Affolter‡ ABSTRACT Gettemans, 2017; Helma et al., 2015; Plückthun, 2015; Sha et al., Developmental biology research would benefit greatly from tools that 2017). These genetically encodable binders, which are based on enable protein function to be regulated, both systematically and in a various protein scaffolds, can be used to block or mask protein ‘ ’ precise spatial and temporal manner, in vivo. In recent years, function. Furthermore, such protein binders can be functionalized functionalized protein binders have emerged as versatile tools that by fusing them to various effector domains with the ultimate goal of can be used to target and manipulate proteins. Such protein binders directly visualizing or regulating the function and interaction of can be based on various scaffolds, such as nanobodies, designed target proteins in living cells or organisms. ankyrin repeat proteins (DARPins) and monobodies, and can be used In the past few years, protein binders have started to be used in to block or perturb protein function in living cells. In this Primer, we developmental biology, and a multitude of novel protein binder- provide an overview of the protein binders that are currently available based tools has been reported. In this Primer, we provide an and highlight recent progress made in applying protein binder-based overview of these tools and discuss the possible applications of tools in developmental and synthetic biology. protein binders in developmental biology research. Furthermore, we highlight the potential and advantages of adding protein binder- KEY WORDS: Protein binder, Nanobody, DARPin, Monobody, based tools to the methodological repertoire of developmental Protein degradation, GFP biologists. Introduction Types of protein binders and their generation Forward and reverse genetic approaches have been key to expanding For many years, antibodies have been the tools of choice for our knowledge of gene function during development (see Housden recognizing a specific protein of interest (POI), and they are still the et al., 2017 and references therein). However, as more and more most widely used reagents for many applications. However, owing to proteins and pathways are becoming associated with developmental their large size and poor folding and stability properties in the processes, and as increasingly complex networks of regulatory cytoplasm of cells, antibodies are most often used in the extracellular interactions emerge (see for example Manning and Toker, 2017), milieu or in fixed tissues, and have proven to be less useful in the the need to study protein function in detail at tissue and single-cell context of a live cell. Over the last decade, however, several classes of scales in living organisms has become greater than ever. This smaller protein-binding scaffolds have been used to generate protein- represents a considerable challenge, in particular when studying specific binders that function when expressed in cells. proteins that regulate general cellular processes or have broad The different classes of peptide-based binders that currently exist expression patterns. The availability of tools that allow protein can be divided into two broad families: (1) those that are based on or function to be regulated more precisely and acutely in a spatial and derived from immunoglobulins, i.e. antibodies and derivatives temporal manner would be extremely helpful and could provide thereof, such as single-chain variable fragments (scFvs) and unprecedented insights into complex developmental processes. nanobodies; and (2) those that are based on non-immunoglobulin, Over the years, several methods have been developed to natural or designed protein scaffolds, such as designed ankyrin manipulate proteins directly in vivo. These include degradation- repeat proteins (DARPins), monobodies, affibodies, anticalins and inducing applications (Banaszynski et al., 2006; Bonger et al., 2011; others (Boxes 1-3, Fig. 1). In the past decade, tremendous progress Chung et al., 2015; Natsume et al., 2016), protein cleavage using has been made in refining these scaffolds for better stability, higher tobacco etch virus (TEV) protease (Harder et al., 2008; Pauli et al., affinity and easier handling (Goldman et al., 2017; Schilling et al., 2008), the ‘anchor-away’ approach (Haruki et al., 2008), the 2014; Schmidt et al., 2016), and libraries of increasing complexity ‘knocksideways’ technique (Robinson et al., 2010) and various have been generated and characterized (Moutel et al., 2016; Tiede dimerization tools that allow protein functions to be assembled in an et al., 2017; Yan et al., 2014). inducible manner (Renicke et al., 2013; van Bergeijk et al., 2015; With the exception of immunoglobulin-based binders, which can Wu et al., 2009), to mention just a few. However, an additional, also be obtained upon immunization (Greenfield, 2014), protein more systematic approach to manipulation of protein function has binders are generally selected using in vitro display techniques recently emerged. This approach utilizes protein binders, which are (Boder and Wittrup, 1997; Samuelson et al., 2002; Zhao et al., small, protein-based affinity reagents that can selectively recognize 2009), with phage display being the most commonly used and bind to a target protein and that are increasingly being used to (Bradbury et al., 2011; Breitling et al., 1991; Romao et al., 2016; study protein function in living cells and organisms (Beghein and Kuhn et al., 2016). Although these techniques use different biological systems, their general principles are shared; they allow large peptide libraries to be screened, the respective protein is Growth and Development, Biozentrum, University of Basel, 4056 Basel, Switzerland. *Present address: IBDM, UMR 7288, Aix-Marseille Universitéand CNRS, 13009 coupled to its encoding DNA sequence, and multiple cycles of Marseille, France. selection can be used to increase binding specificity (Fig. 2). The isolation of protein binders can also be performed commercially and ‡Author for correspondence ([email protected]) on large platforms, and many institutes have established core S.H., 0000-0001-6668-7608; M.A., 0000-0002-5171-0016 facilities to isolate and characterize specific protein binders, making DEVELOPMENT 1 PRIMER Development (2018) 145, dev148874. doi:10.1242/dev.148874 characterization of a novel binder. The latter then has to be Box 1. scFv scaffolds validated with regard to its specificity, efficacy and off-target Conventional antibodies (immunoglobulins, IgGs; see Fig. 1A) have effects, a task that is not always trivial and has to be carefully designed been used extensively in basic research and are indispensable tools for and executed. protein detection. However, conventional IgGs are unsuitable for The second step isto functionalize the binder such that it modifies or intracellular expression for various reasons; the reducing nature of the regulates the target protein in a desired manner. This represents one of intracellular environment hampers disulphide bond formation and thus proper antibody folding, and whole IgG antibodies have a complex the most exciting aspects of protein binders, as there are innumerable structure and a high atomic mass (∼150 kDa). These drawbacks are possibilities for such functionalization. Over the past few years, the partially overcome by connecting the VH and VL domain with a peptide availabilityof awide range of high-affinity protein binders has resulted linker, forming a so-called single-chain variable fragment (scFv; Bird et al., in the development of a versatile repertoire of novel protein binder- 1988; Huston et al., 1988; see Fig. 1B) that retains antigen-binding based techniques and tools (reviewed by Bieli et al., 2016; Böldicke, ∼ capacity. scFvs are relatively small ( 28 kDa) and consist of a single 2017; Helma et al., 2015; Kaiser et al., 2014; Marschall et al., 2015; domain that can be expressed in various host systems, such as bacteria, yeast or higher animals. These advantages make scFvs attractive tools in Plückthun, 2015; Sha et al., 2017). Many of these tools have been medical applications and in biotechnology (Lyon and Stasevich, 2017; validated using binders that recognize specific fluorescent proteins. In Monnier et al., 2013) but their use as intracellular protein binders particular, several binders against green fluorescent protein (GFP) (intrabodies) is restricted because scFvs typically contain two highly have been isolated and characterized (Fridy et al., 2014; Kirchhofer conserved intra-domain disulphide bonds (Williams and Barclay, 1988). et al., 2010; Kubala et al., 2010; Rothbauer et al., 2006; Saerens et al., These bonds influence scFv stability and function (Glockshuber et al., 2005; Brauchle et al., 2014). In addition, several studies have used 1992; Proba et al., 1997), and so only intrinsically stable scFvs fold correctly within a cell and can be utilized as functional intrabodies (Worn binders that recognize specific small tags, such as the 19 amino acid and Pluckthun, 1998). Owing to these limitations, relatively few scFv- (aa) peptide derived from GCN4 used in SunTag (discussed below) based binders have been used to date in developmental biology. (Tanenbaum et al., 2014; Wörn et al., 2000). Nonetheless, using protein engineering (Tanha et al., 2006)
Recommended publications
  • Strategies and Challenges for the Next Generation of Therapeutic Antibodies
    FOCUS ON THERAPEUTIC ANTIBODIES PERSPECTIVES ‘validated targets’, either because prior anti- TIMELINE bodies have clearly shown proof of activity in humans (first-generation approved anti- Strategies and challenges for the bodies on the market for clinically validated targets) or because a vast literature exists next generation of therapeutic on the importance of these targets for the disease mechanism in both in vitro and in vivo pharmacological models (experi- antibodies mental validation; although this does not necessarily equate to clinical validation). Alain Beck, Thierry Wurch, Christian Bailly and Nathalie Corvaia Basically, the strategy consists of develop- ing new generations of antibodies specific Abstract | Antibodies and related products are the fastest growing class of for the same antigens but targeting other therapeutic agents. By analysing the regulatory approvals of IgG-based epitopes and/or triggering different mecha- biotherapeutic agents in the past 10 years, we can gain insights into the successful nisms of action (second- or third-generation strategies used by pharmaceutical companies so far to bring innovative drugs to antibodies, as discussed below) or even the market. Many challenges will have to be faced in the next decade to bring specific for the same epitopes but with only one improved property (‘me better’ antibod- more efficient and affordable antibody-based drugs to the clinic. Here, we ies). This validated approach has a high discuss strategies to select the best therapeutic antigen targets, to optimize the probability of success, but there are many structure of IgG antibodies and to design related or new structures with groups working on this class of target pro- additional functions.
    [Show full text]
  • 2010.01) C07k 14/74 (2006.01
    ( (51) International Patent Classification: su 215 123 (CN). LIN, Yanni; F2, Building B20, Sangt- C07K 19/00 (2006.0 1) C12N 5/0783 (20 10.01) ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu C07K 14/74 (2006.01) A61K 35/1 7 (2015.01) 215123 (CN). ZHENG, Xiaocui; F2, Building B20, Sangt- C12N 15/62 (2006.01) A61P 35/00 (2006.01) ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215123 (CN). KONG, Hongmei; F2, Building B20, Sangt- (21) International Application Number: ian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu PCT/ CN2020/ 1081 16 215123 (CN). WANG, Wenbo; F2, Building B20, Sangtian (22) International Filing Date: Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215 123 10 August 2020 (10.08.2020) (CN). FENG, Aihua; F2, Building B20, Sangtian Street 218, Suzhou Industrial Park, Suzhou, Jiangsu 215 123 (CN). (25) Filing Language: English (74) Agent: J. Z. M. C. PATENT AND TRADEMARK LAW (26) Publication Language: English OFFICE (GENERAL PARTNERSHIP); YU, Mingwei, (30) Priority Data: Room 5022, No. 335, Guo Ding Road, Yang Pu District, 201910746355.8 13 August 2019 (13.08.2019) CN Shanghai 200433 (CN). PCT/ CN20 19/124321 (81) Designated States (unless otherwise indicated, for every 10 December 2019 (10. 12.2019) CN kind of national protection av ailable) . AE, AG, AL, AM, (71) Applicant: CURE GENETICS CO., LTD. [CN/CN]; AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, Biobay A4-5 10, Xinghu Street 218, Suzhou Industrial Park, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, Suzhou, Jiangsu 215 123 (CN).
    [Show full text]
  • EURL ECVAM Recommendation on Non-Animal-Derived Antibodies
    EURL ECVAM Recommendation on Non-Animal-Derived Antibodies EUR 30185 EN Joint Research Centre This publication is a Science for Policy report by the Joint Research Centre (JRC), the European Commission’s science and knowledge service. It aims to provide evidence-based scientific support to the European policymaking process. The scientific output expressed does not imply a policy position of the European Commission. Neither the European Commission nor any person acting on behalf of the Commission is responsible for the use that might be made of this publication. For information on the methodology and quality underlying the data used in this publication for which the source is neither Eurostat nor other Commission services, users should contact the referenced source. EURL ECVAM Recommendations The aim of a EURL ECVAM Recommendation is to provide the views of the EU Reference Laboratory for alternatives to animal testing (EURL ECVAM) on the scientific validity of alternative test methods, to advise on possible applications and implications, and to suggest follow-up activities to promote alternative methods and address knowledge gaps. During the development of its Recommendation, EURL ECVAM typically mandates the EURL ECVAM Scientific Advisory Committee (ESAC) to carry out an independent scientific peer review which is communicated as an ESAC Opinion and Working Group report. In addition, EURL ECVAM consults with other Commission services, EURL ECVAM’s advisory body for Preliminary Assessment of Regulatory Relevance (PARERE), the EURL ECVAM Stakeholder Forum (ESTAF) and with partner organisations of the International Collaboration on Alternative Test Methods (ICATM). Contact information European Commission, Joint Research Centre (JRC), Chemical Safety and Alternative Methods Unit (F3) Address: via E.
    [Show full text]
  • Monobodies and Other Synthetic Binding Proteins for Expanding Protein Science
    Reviews Protein Science DOI 10.1002/pro.3148 Monobodies and Other Synthetic Binding Proteins for Expanding Protein Science Fern Sha,1,4 Gabriel Salzman,1 Ankit Gupta1,2 and Shohei Koide1,2,3* 1Department of Biochemistry and Molecular Biology, The University of Chicago, Chicago, IL 60637, USA 2Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA 3Department of Biochemistry and Molecular Pharmacology New York University School of Medicine, New York, NY 10016, USA 4Present address, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, VA 20148, USA *Correspondence to: Shohei Koide, Perlmutter Cancer Center, New York University Langone Medical Center, New York, NY 10016, USA. E-mail: [email protected] Abstract Synthetic binding proteins are constructed using non-antibody molecular scaffolds. Over the last two decades, in-depth structural and functional analyses of synthetic binding proteins have improved combinatorial library designs and selection strategies, which have resulted in potent platforms that consistently generate binding proteins to diverse targets with affinity and specificity that rival those of antibodies. Favorable attributes of synthetic binding proteins, such as small size, freedom from disulfide bond formation and ease of making fusion proteins, have enabled their unique applications in protein science, cell biology and beyond. Here, we review recent studies that illustrate how synthetic binding proteins are powerful probes that can directly link structure and function,
    [Show full text]
  • WO 2017/013231 Al 26 January 2017 (26.01.2017) P O P C T
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization I International Bureau (10) International Publication Number (43) International Publication Date WO 2017/013231 Al 26 January 2017 (26.01.2017) P O P C T (51) International Patent Classification: BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DK, DM, C07K 14/705 (2006.01) C07K 16/00 (2006.01) DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JP, KE, KG, KN, KP, KR, (21) International Application Number: KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, PCT/EP20 16/067468 MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, (22) International Filing Date: PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, 2 1 July 20 16 (21 .07.2016) SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW. (25) Filing Language: English (84) Designated States (unless otherwise indicated, for every (26) Publication Language: English kind of regional protection available): ARIPO (BW, GH, (30) Priority Data: GM, KE, LR, LS, MW, MZ, NA, RW, SD, SL, ST, SZ, 62/194,882 2 1 July 2015 (21.07.2015) TZ, UG, ZM, ZW), Eurasian (AM, AZ, BY, KG, KZ, RU, 62/364,414 20 July 2016 (20.07.2016) TJ, TM), European (AL, AT, BE, BG, CH, CY, CZ, DE, DK, EE, ES, FI, FR, GB, GR, HR, HU, IE, IS, IT, LT, LU, (72) Inventors; and LV, MC, MK, MT, NL, NO, PL, PT, RO, RS, SE, SI, SK, (71) Applicants : XIANG, Sue D.
    [Show full text]
  • Protein Scaffolds Proteingerüste Structures Protéiques
    (19) TZZ _ _T (11) EP 2 215 246 B1 (12) EUROPEAN PATENT SPECIFICATION (45) Date of publication and mention (51) Int Cl.: of the grant of the patent: C12N 15/10 (2006.01) C12N 15/62 (2006.01) 07.01.2015 Bulletin 2015/02 C07K 14/78 (2006.01) C40B 40/10 (2006.01) (21) Application number: 08845766.8 (86) International application number: PCT/US2008/012398 (22) Date of filing: 31.10.2008 (87) International publication number: WO 2009/058379 (07.05.2009 Gazette 2009/19) (54) PROTEIN SCAFFOLDS PROTEINGERÜSTE STRUCTURES PROTÉIQUES (84) Designated Contracting States: US-B1- 6 482 410 US-B1- 6 818 418 AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT • KOIDE A ET AL: "The fibronectin type III domain RO SE SI SK TR as a scaffold for novel binding proteins", JOURNAL OF MOLECULAR BIOLOGY, LONDON, (30) Priority: 31.10.2007 US 984209 P GB, vol. 284, no. 4, 11 December 1998 (1998-12-11), pages 1141-1151, XP004455886, (43) Date of publication of application: ISSN: 0022-2836, DOI: DOI:10.1006/JMBI. 11.08.2010 Bulletin 2010/32 1998.2238 • BATORI V ET AL: "Exploring the potential of the (73) Proprietor: MedImmune, LLC monobody scaffold:effects of loop elongation on Gaithersburg, MD 20878 (US) the stability of a fibronectin type III domain", PROTEIN ENGINEERING, OXFORD UNIVERSITY (72) Inventors: PRESS, SURREY, GB, vol. 15, no. 12, 1 January • WU, Herren 2002 (2002-01-01), pages 1015-1020, Boyds,MD 20841 (US) XP002996047, ISSN: 0269-2139, DOI: DOI: • BACA, Manuel 10.1093/PROTEIN/15.12.1015 Gaithersburg,MD 20878 (US) • KOIDE AKIKO ET AL: "Monobodies: antibody •SWERS,Jeffrey mimics based on the scaffold of the fibronectin Rockville,MD 20852 (US) type III domain", METHODS IN MOLECULAR • CHACKO, Benoy BIOLOGY, HUMANA PRESS INC, NJ, US, vol.
    [Show full text]
  • Highly Potent Anti-SARS-Cov-2 Multi-Darpin Therapeutic Candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L
    bioRxiv preprint doi: https://doi.org/10.1101/2020.08.25.256339; this version posted August 26, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. Highly potent anti-SARS-CoV-2 multi-DARPin therapeutic candidates Marcel Walser1,*, Sylvia Rothenberger2,3,*, Daniel L. Hurdiss4,5,*, Anja Schlegel1, Valérie Calabro1, Simon Fontaine1, Denis Villemagne1, Maria Paladino1, Tanja Hospodarsch1, Alexandra Neculcea1, Andreas Cornelius1, Patricia Schildknecht1, Mirela Matzner1, Martin Hänggi1, Marco Franchini1, Yvonne 5 Kaufmann1, Iris Schlegel1,, Chloe Iss1, Thamar Loser1, Susanne Mangold1, Christel Herzog1, Dieter Schiegg1, Christian Reichen1, Filip Radom1, Andreas Bosshart1, Andreas Lehmann1, Micha A. Haeuptle1, Alexander Zürcher1, Toni Vagt1, Gabriel Sigrist1, Marcel Straumann 1, Karl Proba1, Niina Veitonmäki1, Keith M. Dawson1, Christof Zitt1, Jennifer Mayor2,3, Sarah Ryter2, Heyrhyoung Lyoo4, Chunyan Wang4, Wentao Li4, Ieva Drulyte6, H. Kaspar Binz7, Leon de Waal8, Koert J. Stittelaar8, Seth Lewis1, Daniel 10 Steiner1, Frank J.M. van Kuppeveld4, Olivier Engler2, Berend-Jan Bosch4, Michael T. Stumpp1,9, Patrick Amstutz 1 *These authors contributed equally to this work 1Molecular Partners AG, Wagistrasse 14, 8952 Zurich-Schlieren, Switzerland 15 2Spiez Laboratory, Austrasse, 3700 Spiez, Switzerland 3Institute of Microbiology, University Hospital Center
    [Show full text]
  • Understanding Ubiquitin Recognition and Generating Affinity Reagents
    Ubiquitin Engineering: Understanding Ubiquitin Recognition and Generating Affinity Reagents by Isabel Leung A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Molecular Genetics University of Toronto © Copyright by Isabel Leung 2017 Abstract Ubiquitin Engineering: Understanding Ubiquitin Recognition and Generating Affinity Reagents Isabel Leung Doctor of Philosophy Department of Molecular Genetics University of Toronto 2016 Protein-protein interactions are necessary for virtually all biological processes. There have been tremendous efforts to document the diversity of molecular recognition, and to understand how molecular recognition occurs. The understanding of molecular interaction has also served as the foundation for designing novel protein interactions for use in therapeutics, diagnostics and basic sciences. An attractive system for studying protein-protein interactions is the ubiquitin (Ub) system. Ub is a protein modifier that is combinatorially ligated onto substrate proteins to influence substrate turnover and function. Ub uses a common surface to interact with more than 1000 proteins and plays pivotal roles in cell physiology. Despite the substantial structural information on Ub mediated interactions, there is no clear understanding of how individual Ub residues contribute to Ub’s broad scope of interactions. To address this question, I used affinity enhanced Ub variants (Ubvs) as proxies of native Ub in saturation scanning. Using saturation scanning, I studied the interactions between Ubvs and two Ub specific proteases (USP), USP2 and USP21, and elucidated a common functional epitope that is critical for USP recognition. The functional epitope recognizes USP residues that are conserved among the human USP family, suggesting it may make functional contributions in many other USP interactions.
    [Show full text]
  • (12) United States Patent (10) Patent No.: US 9,499,622 B2 Cheong Et Al
    USOO9499622B2 (12) United States Patent (10) Patent No.: US 9,499,622 B2 Cheong et al. (45) Date of Patent: Nov. 22, 2016 (54) ANTI-EGFR/ANTI-HER2 BISPECIFIC 5,821,337 A 10, 1998 Carter et al. ANTIBODIES WITH ANT-EGFR DARPNS 6,165,464 A 12/2000 Hudziak et al. 7,417,130 B2 8/2008 Stumpp et al. 8,071,730 B2 12/2011 Goetsch et al. (71) Applicant: Samsung Electronics Co., Ltd., 8, 110,653 B2 2/2012 Stumpp et al. Suwon-si, Gyeonggi-do (KR) 8,329,873 B2 12/2012 Adams et al. 8,524,244 B2 9/2013 Camphausen et al. (72) Inventors: Kwang Ho Cheong, Seoul (KR): 9,234,028 B2 1/2016 Camphausen et al. Seung Hyun Lee, Suwon-si (KR): 9,359.440 B2 * 6/2016 Cheong ................ CO7K 14,705 Powei Lin, Hwaseong-si (KR); Saet 2009/0010840 A1 1/2009 Adams et al. Byoul Lee, Seoul (KR); Jae Woong 2010/0178298 A1 7/2010 Lindhofer 2012fOO2O966 A1* 1/2012 Barbas, III ........... CO7K 14,515 Hwang, Seoul (KR) 424,134.1 2012/0156191 A1 6/2012 Goetsch et al. (73) Assignee: SAMSUNGELECTRONICS CO., 2012,0277143 A1 1 1/2012 Jacobs et al. LTD., Suwon-Si (KR) 2013, OO17200 A1 1/2013 Scheer et al. 2015.OO30596 A1* 1/2015 Cheong ................ CO7K 14,705 (*) Notice: Subject to any disclaimer, the term of this 424,134.1 patent is extended or adjusted under 35 U.S.C. 154(b) by 0 days. FOREIGN PATENT DOCUMENTS EP 1332.209 B2 11/2009 (21) Appl.
    [Show full text]
  • University of Copenhagen, DK-2200 København N, Denmark * Correspondence: [email protected]; Tel.: +45-2988-1134 † These Authors Contributed Equally to This Work
    Toxin Neutralization Using Alternative Binding Proteins Jenkins, Timothy Patrick; Fryer, Thomas; Dehli, Rasmus Ibsen; Jürgensen, Jonas Arnold; Fuglsang-Madsen, Albert; Føns, Sofie; Laustsen, Andreas Hougaard Published in: Toxins DOI: 10.3390/toxins11010053 Publication date: 2019 Document version Publisher's PDF, also known as Version of record Citation for published version (APA): Jenkins, T. P., Fryer, T., Dehli, R. I., Jürgensen, J. A., Fuglsang-Madsen, A., Føns, S., & Laustsen, A. H. (2019). Toxin Neutralization Using Alternative Binding Proteins. Toxins, 11(1). https://doi.org/10.3390/toxins11010053 Download date: 09. apr.. 2020 toxins Review Toxin Neutralization Using Alternative Binding Proteins Timothy Patrick Jenkins 1,† , Thomas Fryer 2,† , Rasmus Ibsen Dehli 3, Jonas Arnold Jürgensen 3, Albert Fuglsang-Madsen 3,4, Sofie Føns 3 and Andreas Hougaard Laustsen 3,* 1 Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK; [email protected] 2 Department of Biochemistry, University of Cambridge, Cambridge CB3 0ES, UK; [email protected] 3 Department of Biotechnology and Biomedicine, Technical University of Denmark, DK-2800 Kongens Lyngby, Denmark; [email protected] (R.I.D.); [email protected] (J.A.J.); [email protected] (A.F.-M.); sofi[email protected] (S.F.) 4 Department of Biology, University of Copenhagen, DK-2200 København N, Denmark * Correspondence: [email protected]; Tel.: +45-2988-1134 † These authors contributed equally to this work. Received: 15 December 2018; Accepted: 12 January 2019; Published: 17 January 2019 Abstract: Animal toxins present a major threat to human health worldwide, predominantly through snakebite envenomings, which are responsible for over 100,000 deaths each year.
    [Show full text]
  • (10) Patent No.: US 9034822 B2
    USOO9034822B2 (12) United States Patent (10) Patent No.: US 9,034,822 B2 Van Ryn et al. (45) Date of Patent: *May 19, 2015 (54) METHODS OF USING ANTIBODIES DURING (56) References Cited ANTCOAGULANT THERAPY OF DABGATRAN AND/OR RELATED U.S. PATENT DOCUMENTS COMPOUNDS 6,440,417 B1 8/2002 Thibaudeau et al. 6,469,039 B1 10/2002 Hauel et al. (71) Applicants: Joanne Van Ryn, Warthausen (DE); 8,486,398 B2* 7/2013 Van Ryn et al. ........... 424,133.1 2004/OO97547 A1 5, 2004 Taveras et al. John Edward Park, Warthausen (DE): 2011/0206656 A1 8/2011 Van Ryn et al. Norbert Hauel, Schemmerhofen (DE): 2012fOO27780 A1 2/2012 Van Rynet al. Ulrich Kunz, Biberach an der Riss (DE); Tobias Litzenburger, Mittelbiberach FOREIGN PATENT DOCUMENTS (DE); Keith Canada, Southbury, CT CA 2 277 949 8, 1998 (US); Sanjaya Singh, Sandy Hook, CT CA 2277949 A1 * 8, 1998 (US); Alisa Waterman, Weston, CT WO WO-9837O75 8, 1998 (US) WO WO-2011 O23.653 3, 2011 WO WO-2011 089183 T 2011 (72) Inventors: Joanne Van Ryn, Warthausen (DE); John Edward Park, Warthausen (DE): OTHER PUBLICATIONS Norbert Hauel, Schemmerhofen (DE): Schlaeppi et al., Eur J Biochem. Mar. 10, 1990:188(2):463-70.* Ulrich Kunz, Biberach an der Riss (DE); Herion et al., Blood. May 1985:65(5):1201-7.* Tobias Litzenburger, Mittelbiberach Colburn, W. A., “Specific antibodies and fab fragments to alter the (DE); Keith Canada, Southbury, CT pharmacokinetics and reverse the pharmacologic/toxicologic effects (US); Sanjaya Singh, Sandy Hook, CT of drugs.” Drug Metabolism Reviews, 1980, vol.
    [Show full text]
  • Single-Round, Multiplexed Antibody Mimetic Design Through Mrna Display** C
    Angewandte Chemie DOI: 10.1002/anie.201207005 Ligand Design Single-Round, Multiplexed Antibody Mimetic Design through mRNA Display** C. Anders Olson, Jeff Nie, Jonathan Diep, Ibrahim Al-Shyoukh, Terry T. Takahashi, Laith Q. Al- Mawsawi, Jennifer M. Bolin, Angela L. Elwell, Scott Swanson, Ron Stewart, James A. Thomson, H. Tom Soh, Richard W. Roberts, and Ren Sun* There is a pressing need for new technologies to generate fold enrichment. Our results also demonstrate that highly robust, renewable recognition tools against the entire human functional binders (K 100 nm or better) are present with D proteome.[1] Hybridoma-based monoclonal antibodies,[2] the a frequency of > 1 in 109 in our library. standard for protein reagents, are undesirable for this task To begin, we needed to devise an appropriate selection because of the number of animals, amount of target, time, and protocol and library creation format. Typically, in vitro effort required to generate each reagent. Here, we developed selections require multiple rounds of modest sequential a unified approach to solve this problem by integrating four enrichment, followed by small-scale sequencing of functional distinct technologies: 1) a combinatorial protein library based clones (Figure 1A). Indeed, the need to generate a target- on the 10th fibronectin type III domain of human fibronectin specific library at each round provides a significant limitation (10Fn3),[3] 2) protein library display by mRNA display,[4] towards parallelizing and accelerating selections. In contrast, 3) selection by continuous-flow magnetic separation for identification of ligands after a single round of CFMS (CFMS),[5] and 4) sequence analysis by high throughput mRNA display (Figure 1A,B), only a single naive library pool sequencing (HTS).[6] Next generation sequencing has revolu- must be synthesized for any number of targets, drastically tionized many fields of biology, and is increasingly being used decreasing the effort needed for ligand discovery.
    [Show full text]